Ndubuisi N. N, Herbert A. O. Therapeutic Drug Monitoring in Pediatric Practice: A Critical Appraisal. Biomed Pharmacol J 2014;7(1)
Manuscript received on :
Manuscript accepted on :
Published online on: 23-12-2015
How to Cite    |   Publication History
Views Views: (Visited 983 times, 1 visits today)   Downloads PDF Downloads: 634

N. Nwobodo Ndubuisi1 and A. Obu Herbert2

1Department of Pharmacology and Therapeutics, Faculty of Clinical Medicine, Ebonyi State University, Abakaliki, Nigeria.

2Department of Pediatrics, College of Medicine, University of Nigeria.

Corresponding Author E-mail: nnwobodo@yahoo.com

DOI : https://dx.doi.org/10.13005/bpj/479

Abstract

The criteria for drug monitoring in children are almost similar as applicable in adults, though certain factors need to be taken into consideration. The dramatic pace of change in other areas of clinical therapeutics has not reflected significantly in the specialty of pediatrics, accounting for the poor development of therapeutic drug monitoring in children. Major drug interactions in the pediatric population have been revealed. CYP3A4 hepatic microsomal enzyme plays a major role in these drug interactions.The continuing education and awareness of these interactions among healthcare practitioners is critical in optimizing effectiveness and minimizing toxicity. Therapeutic drug monitoring is primarily indicated for drugs with narrow therapeutic margin; though certain category of patients may still manifest evidence of toxicity, despite drug concentration being within the therapeutic range. The use of pharmacodynamic data in synergy with therapeutic drug monitoring represents the most viable approach to individualized therapy. Pediatrics is at the epicenter of the emerging discoveries in the field of genomic medicine. The relevance of therapeutic drug monitoring as a global therapeutic index encompassing pharmacokinetics, pharmacodynamics, pharmacogenomics and drug interactions can never be overemphasized. In conclusion, the prospects of clinical pharmacogenomics as therapeutic drug monitoring for the future in pediatric practice is quite promising.

Keywords

Appraisal; Clinical pharmacogenomics; Drug disposition; Drug interaction; pharmacodynamics; Pharmacokinetics; Therapeutic drug monitoring

Download this article as: 
Copy the following to cite this article:

Ndubuisi N. N, Herbert A. O. Therapeutic Drug Monitoring in Pediatric Practice: A Critical Appraisal. Biomed Pharmacol J 2014;7(1)

Copy the following to cite this URL:

Ndubuisi N. N, Herbert A. O. Therapeutic Drug Monitoring in Pediatric Practice: A Critical Appraisal. Biomed Pharmacol J 2014;7(1). Available from: http://biomedpharmajournal.org/?p=2929

Introduction

Therapeutic drug monitoring can be defined as measurement of drug concentrations in biologic matrix with a view to assessing correlation with patient’s clinical condition and the need for dose adjustment. The criteria for drug monitoring in children are almost the same as applicable in adults, though certain factors should be taken into consideration1. It has been shown that children aged 9 years and below receive approximately 12% of all drugs prescribed in the United State of America alone2. Pharmacokinetic and pharmacodynamic behaviour differ significantly in the pediatric age group compared to the normal adult population. Parental compliance in administering drugs at the appropriate time interval may further accentuate patient non-compliance in the pediatric population. The dramatic pace of change in other areas of therapeutics has not reflected in the specialty of pediatrics accounting for the poor development of therapeutic drug monitoring in children3. It is therefore, no wonder that infant, toddlers and children being denied access to benefits of modern drug therapy are referred to as “therapeutic orphans”. This paper examines the fundamental issues underlying therapeutic drug monitoring in pediatric practice with a view to optimizing patient care.

Materials and Methods

An advanced search of literature using pubmed central, medline and embase was carried out with a view of accessing peer reviewed full journal articles, abstracts, reviews, comments, letters to editors, project reports, dissertations, theses and books relevant to the subject matter.. The keywords used in the search were as follows: appraisal, drug disposition, drug interaction, pharmacodynamics, pharmacogenetics, pharmacogenomics, pharmacokinetics and therapeutic drug monitoring.

Drug Interactions

Drug interactions generally refer to effects of concomitant administration of a drug with other drugs (drug-drug interaction) as well as drugs with food (food-drug interaction) or other substances which results to a clinically measurable modification in either magnitude or duration of action of the index drug. Changes in drug disposition brought about by a particular drug can alter the pharmacokinetics of another drug. The clinical consequences of these interactions may manifest as sub-therapeutic effect due to reduced serum drug concentration or increased adverse effects due to elevated level of serum drug concentration. This clearly underscores the need for drug monitoring to ensure that requisite target concentration is achieved.

Drug-Drug Interaction

A significant number of adverse events in hospitalized patients as evidenced by epidemiologic studies is accounted for by drug-drug interactions4,5. A study revealed variety of major drug interactions in the pediatric population, highlighting cases of first significance rate interaction with rapid onset6. Electrolyte changes particularly potassium loss induced by loop diuretics results to hypokalemia potentiating digoxin toxicity. Moreover, at low serum potassium level, tubular secretion of digoxin is inhibited, further increasing digoxin serum concentration and prolonging its elimination half-life with consequent risk of cardiac arrhythmia. Concomitant administration of the non-sedating antihistamine terfenadine with macrolide antibiotics should be avoided due to risk of cardiotoxicity. There is need for caution in the co-administration of loop diuretics and aminoglycoside antibiotics due to synergistic potentiation of oxotoxicity; although dose-dependent toxicity may still manifest in the course of administering drugs individually7. Predictable drug interaction occurs during concomitant administration of azole antifungals  such as ketoconazole, itraconazole, voriconazole with barbiturates leading to their increased metabolism and sub-therapeutic serum concentration mediated by induction of microsomal liver enzymes by barbiturates. Other classes of drugs in which increased metabolism is reported following concomitant administration with barbiturates include: beta-adrenoceptor blockers, calcium channel blockers, antidepressants and corticosteroids. Rifampin is a strong inducer of hepatic microsomal drug metabolizing enzyme and co-administration with drugs such as dexamethasone, theophylline, paracetamol and tolbutamide will result to increased metabolism and reduced therapeutic effects of these drugs. Increase in the metabolism of paracetamol induced by rifampin results to accumulation of metabolites which are hepatotoxic8.

Food-Drug Interactions

Food-drug interaction is the effect produced when certain foods or beverages are taken concomitantly with drugs. Food-drug interactions alter the pharmacokinetics or pharmacodynamics of a drug or nutritional element. Regrettably, consensus toward specific drug–nutrient interactions, standardized management approaches and properly designed studies on the epidemiology of food-drug interactions are still lacking. The continuing education and awareness of these interactions among healthcare practitioners is critical in optimizing effectiveness and minimizing toxicity9. Altered bioavailability of a drug such as complex formation with metal ion, partitioning in dietary fat or adsorption of drug in insoluble dietary component may occur due to direct interaction of food with a drug. The microsomal hepatic drug metabolizing enzymes particularly CYP3A4 play a key role in food-drug interaction10. Elevated serum concentration of certain drugs by more than five-fold following ingestion of grape fruit has been reported and linked to enzymatic inhibition of selective microsomal drug metabolizing enzymes11. Grapefruit juice has no effect on drug pharmacokinetic parameters after intravenous administration but causes significant rise in drug bioavailability after ingestion, suggesting that it has no effect on liver CYP3A4 but significantly inhibits intestinal CYP3A412,13.

Bergamothin is the major furanocoumarin found in grape fruit responsible for drug interaction, exhibiting both concentration and time-dependent in vitro  inactivation of cytochrome P450 microsomal enzymes; furthermore, its metabolite also inhibits CYP1B1 and CYP3A414,15.

Drug interaction with grapefruit juice is influenced by the time of ingestion. It has been reported that  12 hours after intake of grapefruit juice, the bioavailability of lovastatin doubled16, though a clinically significant interaction did not occur after an interval of 24 hours17. A study revealed that grapefruit juice did not have any significant effect on maximal plasma concentration of digoxin, a substrate of P-glycoprotein18. Orange juice significantly reduced area under the curve, AUC of celiprolol by 83% and the mean peak plasma concentration by 89%19. The AUC of felodipine was increased by sour orange to 76% compared to 93% by grapefruit juice20.

Drugs Routinely Monitored

Therapeutic drug monitoring is primarily indicated for drugs with narrow therapeutic margin in which the serum drug concentration that results to adverse effect is quite close to the concentration required to achieve beneficial therapeutic effect. It is pertinent to note, however, that certain patients may still exhibit adverse effects, even in situation where drug concentration is within therapeutic margin due to variations in individual pharmacokinetic and pharmacodynamic indices.

Digoxin is a cardiac glycoside derived from Digitalis lanata. Digitalis can be accurately monitored using HPLC combined with tandem mass spectrometry. Digoxin immunoassays have the advantage of rapid turnaround time and automation, but subject to interference by endogenous digoxin-like immunoreactive substances (DLIS) due to their structural  similarity. Digoxin may accumulate in smaller amounts in immature infants due to diminution in total body fat seen in preterms. Digoxin is weakly protein bound as compared to DLIS which are strongly protein bound, hence free drug concentration measurement of digoxin is preferable in order to eliminate interference. A study indicated that toxic effects of digoxin appear at concentrations from 1.2ng/mL, whereas the therapeutic effects occurred within concentration range of 0.5 to 0.9 ng/mL21. A study reported poor correlation between DLIS concentration and patient age, total bilirubin and serum creatinine level22. Moreover, concentrations of DLIS in maternal blood may be significantly decreased relative to cord blood.

Specific clinical indications for monitoring of anticonvulsant therapy in pediatric patients include determinantion of baseline effective concentrations, evaluating cases of toxicity, lack of efficacy and non-compliance. It has since been shown that dosing of anticonvulsant drugs based solely on mg/kg body weight was not effective23,24. A study reported that despite drug concentration below optimal therapeutic interval established at the time, a number of epileptic patients undergoing treatment remained seizure free25. Measurement of free phenytoin levels in suspected cases of toxicity where total serum phenytoin is within the optimal therapeutic range may be necessary as phenytoin is highly protein bond (90%). Carbamazepine is one of the most commonly used anticonvulsants approved for children over six years. Immunoassay method widely used for measuring carbamazepine concentration in blood, is subject to interferences due to cross-reactivity with carbamazepine metabolites and other structurally similar compounds26. Phenobarbital is a sedative hypnotic effective in the treatment of epilepsy with the exception of absence seizures, though not currently recommended as first or second line drug for seizure control in children. Toxic effects including alteration in level of consciousness, shallow breathing, bradycardia and renal failure occur with overdose. Cross-reactive interferences with amobarbital, butobarbital, secobarbital and phenytoin have been reported following phenobarbital immunoassay27.

Increased incidence of toxicity occurs in asthmatic children at theophylline plasma concentration above 20mcg/mL. Serum concentration 10 to 20 mcg/mL is effective in relieving asthmatic attack in children. CYP1A2 microsomal enzyme is responsible for metabolism of theophylline which is reported to be faster in females compared to males. A study has shown that steady state serum concentration of theophylline was reduced by 24.5% while the theophylline clearance increased by 51.1% in children exposed to passive smoking28. A twofold reduction in half life of theophylline was reported in smokers relative to non-smokers29.

Increased risk of otoxicity and nephrotoxicity seen in aminoglycosides such as gentamicin and tobramycin is associated with sustained peak concentration above 12-15µg/ml and/or trough levels exceeding 2mcg/ml. The clearance of aminoglycosides is increased in children as compared to adults; and patients with fever exhibited lower plasma concenetration and shorter half-life30. Glomerular filtration rate (GFR) on which clearance of aminoglycoside depends is drastically lowered in neonates particularly premature newborns. Prolonged half-life of aminoglycosides in neonates may be accounted for by the increase in volume of distribution,Vd of aminoglycosides in neonates. Hence, increase in Vd and reduced clearance of gentamicin have been observed in neonates31. Vancomycin which is excreted in urine unchanged is frequently monitored due to its low therapeutic index, complicating therapy with combined risk of ototoxicity and nephrotoxicity32. Monitoring of trough and peak concentrations of vancomycin and their ranges is quoted in literature33. However, a conservative range of 20-40µg/mL for peak concentration and 5–15µg/mL for the trough is recommended for infants34. Trough concentrations above 30ng/mL and 80 to 100ng/mL may be associated with increased risk of nephrotoxicity and ototoxicity respectively. The decrease in clearance of most beta lactam antibiotics is as a result of reduced renal clearance in neonates35.

The lack of adequate viral suppression in the absence of therapeutic drug monitoring of anti-retroviral drugs has been shown by various studies. The incidence of inter-patient variability and drug-drug interactions in pediatric population is one of the major indications for therapeutic drug monitoring of antiretroviral drugs. Available evidence  is suggestive that both non-nucleoside reverse transcriptase inhibitors, NNRTIs such as nevirapine, delavirdine, efavirenz and protease inhibitors, PI such as saquinavir, indinavir, atazanavir, lopinavir, ritonavir, neltinavir are good candidates for therapeutic drug monitoring; while nucleoside reverse transcriptase inhibitors, NRTIs such as zidovudine, lamivudine, stavudine, zalcitabine and  didanosine  are not. The simultaneous measurement of any combination of antiretroviral drugs using tandem mass spectrometry has facilitated assessment of both compliance and optimization of dosage regimens in children36,37.

Future Propects

The vast genetically determined variations in drug response makes even more difficult the search for optimized pharmacotherapy38. The use of pharmacodynamic data in synergy with therapeutic drug monitoring represents the most viable approach to individualized therapy. The identification of genotype as aid to therapeutic drug monitoring is a very promising prospect39. Notwithstanding, knowledge of measurement of serum drug concentration followed by appropriate adjustment, still remains inevitable as awareness of metabolizer status may not be sufficient to allow for prediction of serum drug concentration measurement. The drastic reduction in the cost of genotyping and more importantly next generation sequencing techniques, following the successful completion of the Human Genome Project have led to insights into gene regulation and complex interplay of factors responsible for normal development. The emerging fields of clinical pharmacogenomics and practice of personalized medicine are among the most tangible outcome of the Human Genome Project40-42. Pharmacogenomic biomarkers are useful adjuncts to facilitate practice of personalized medicine. Pediatrics is at the epicenter of the emerging discoveries in the field of genomic medicine. Notwithstanding the daunting challenges of translating genomic knowledge into improved patient care, pediatricians and their patients are favourably disposed towards benefiting maximally from this genomic revolution43.

In conclusion, the relevance of therapeutic drug monitoring in pediatrics encompassing pharmacokinetics, pharmacodynamics, pharmacogenomics, drug interactions, selection of appropriate drugs and techniques for monitoring can never be overemphasized. The prospects of clinical pharmacogenomics as therapeutic drug monitoring for the future in pediatric practice is quite promising.

References

  1. Christians U, First WR, Benet LZ. Recommendations for bio-equivalence testing of cyclosporine generics revisited. Ther Drug Monit 2000; 22: 330–345.
  2. Loebstein R, Koven G. Clinical Pharmacology and therapeutic drug monitoring in neonates and children. Pediatr Rev 1998; 19: 423–428.
  3. Macleod S. Therapeutic drug monitoring in pediatrics: how do children differ? Ther Drug Monit 2010; 32(3): 253–256.
  4. Leape LL, Brennan TA, Laird N, Lawthers AG, Localio AR, Varnes BA, Herbert L, Newhouse JP, Weiler PC, Hiatt H. The nature of adverse events in hospitalized patients: Results from the Harvard Medical Practice Study II. N Engl J Med 1991; 324: 377–384.
  5. Leape LL, Bates DW, Cullen DJ, Cooper J, Demonaco HJ, Gallivan T, Hallisey R, Ives J, Laird N, Laffel G, Nemeskal R, Peterson LA, Porter K, Servi D, Shea BF, Small SD, Sweetzer BJ, Thompson BT, Viet MV, Bates D, Diaz-Hojnowski P, Petrycki S, Cotugno M, Patterson H, Hickey M, Kleefield S, Kinneally E, Clapp MD, Hackman JR, Edmondson A. Systems analysis of adverse drug events. JAMA 1995; 274: 35–43.
  6. Qorraj–Bytyqi H., Hoxha R,Krasniqi S, Bahtiri E, Krasniqi V. The incidence and clinical relevance of drug interactions in pediatrics. J Pharmacol Phamacother 2012;    3(4): 304–307.
  7. Bates DE, Beaumont SJ, Baylis BW. Ototoxicity induced by gentamicin and furosemide. Ann Pharmacother 2002; 36: 446–451.
  8. Stephenson I, Qualie M, Wisella MJ. Hepatic failure and encephalopathy attributed to an interaction between acetaminonphen and rifampin. Am J Gastroenterol 2001; 96: 1310–1311.
  9. Maka DA, Murphy LK. Drug-nutrient interactions: a review. AACN Clin Issues 2000; 11(4): 580-589.
  10. Paine MF, Widmer WW, Hart HL, Pusek SN, Beavers KL, Criss AB, Brown SS, Thomas BF, Watkins PB. A furanocoumarin-free grapefruit juice establishes furanocoumarins as the mediators of the grapefruit juice–felodipine interaction. Am J Clin Nutr 2006;   83: 1097 – 1105.
  11. Fujita K. Food drug interactions via human cytochrome P450 3A4 (CYP3A4). Drug Metabol Drug Interact 2004; 20: 195–217.
  12. Uno T, Ohkubo I, Sugawara K, Higashiyama A, Motomura S, Ishizaki T. Effects of grapefruit juice on stereoselective disposition of nicardipine in humans: Evidence for dominant presystematic elimination at the gut site. Eur J Clin Pharmacol 2000; 56: 643–649.
  13. Daha A, Altman H. Food-drug interaction: grape fruit juice augments bioavailability-mechanism, extent and relevance. Eur J Clin Nutr 2004; 58: 1-9.     
  14. 14. He K, Iyer KR, Hayes RN, Sinz MW, Woolf TF, Hollenberg PF. Inactivation of cytochrome P450 3A4 by bergamottin, a component of grape fruit juice. Chem Res Toxicol 1998; 11: 252–259.
  15. Girennavar B, Poulose SM, Jayaprakasha GK, Bhat NG, Paul BS. Furocoumarins from grapefruit juice and their effect on human CYP3A4 and CYP1B1 isoenzymes. Bioorg Med Chem 2006, 14; 2602–2612.
  16. Rogers JD, Zhao J, Liu L, Amin RD, Gagliano KD, Porras AG. Grapefruit juice has minimal effect on plasma concentrations of lovastatin derived 3-hydroxy 3-methyl glutaryl coenzyme  A reductase inhibitors. Clin Pharmacol Ther 1999; 66: 58–366.
  17. Lilja J J, Kiristo KT, Neuvoen PJ. Duration of effect of grapefruit juice on pharmacokinetic of the CYP3A4 substrate simvastatin. Pharmacol Ther 2000; 68: 309–390.
  18. Becoquemont L, Verstuyft C, Kerb R, Brinkmann U, Lebot M, Patrice Jaillon P, Funck-Brentano C. Effect of grapefruit juice on digoxin pharmacokinetics in humans. Clin Pharmacol Ther 2001; 70: 311–316.
  19. Lilja JJ, Juntti–Patinen L, Neuvonen PJ. Orange juice substantially reduced the bioavailability of the beta–adreneragic blocking agent celiprolol. Clin Pharmacol Ther 2004; 75: 184–190.
  20. Mahatra S, Bailey DG, Paine MF, Watkins PB, Seville orange juice–felodipine interaction: comparison with dilute grapefruit and involvement of furocoumarins. Clin Pharmacol Ther 2001; 69: 14-23.
  21. Adams KF, Patterson JH, Gattis WA, O’Connor CM, Lee CR, Schwartz TA, Gheorghiade M. Relationship of serum digoxin concentrations to mortality and morbidity in women in the digitalis investigation group trial: a retrospective study. J Am Coll Cardiol 2005; 46: 497 – 504
  22. Chicella M, Branim B, Lee KR, Phelps SJ. Comparison of microparticle enzyme and fluorescence polarization immunoassays in pediatric patients not receiving digoxin. Ther Drug Monit 1998; 20: 347–351.
  23. Pippenger CE, Gillen HW. Gas chromatographic analysis for anticonvulsant drugs in biologic fluids. Clin Chem 1969; 15: 582–590.
  24. Kupferberg HJ. Quantitative estimation of diphenylhydantoin, primidone and phenobarbital in plasma by gas-liquid chromatography. Clin Chim Acta 1970;29; 282 – 288.
  25. Fieldman RG, Pippenger CE. The relation of anticonvulsant drug levels to complete seizure control. J Clin Pharmacol 1976; 16: 51–59.
  26. Shen S, Elin RJ, Soldin SJ. Characterization of cross-reactivity by carbamazepine 10, 11-epoxide with carbamazepine assay. Clin Biochem 2001; 34: 157–158.
  27. Ammann H, Vinet B. Accuracy, precision and interferences of three modified EMIT procedures for determining serum phenobarbital, urine morphine and urine metabolites with a cobas-fara. Clin Chem 1991; 37: 2139-2141.
  28. Lee B L, Benowitz NL, Jacob P. Cigarette abstinence, nicotine gum and theophylline disposition. Ann Inter Med 1987; 1964(4): 553–555.
  29. Zevin S, Benowitz NL. Drug Interactions with tobacco smoking: an update. Clin Pharmacokinet 1999; 36: 425–438.
  30. Siber GR, Echeverria P, Smith AL, Paisley JW, Smith DH. Pharmacokinetics of gentamicin in children and adults. J Infect Dis 1975; 132: 637–651.
  31. Williams BS, Ransom JL, Gal P, Carlos RQ. Gentamicin pharmacokinetics in neonates with patent ductus arteriosus. Crit Care Med 1997; 25: 272-273.
  32. Duffull SB, Begg EJ. Vancomycin toxicity: what is the evidence for dose dependence? Adverse Drug React Toxicol Rev 1994; 13: 103–114.
  33. Begg EJ, Barclay ML, Kirkpatrick C. The therapeutic monitoring of antimicrobial agents. Br J Clin Pharamcol 1999; 47: 23–30 [Review].
  34. de Hoog M, Schoemaller RC, Mootor JW, van der Anker JN. Vancomycin population pharmacokinetics in neonates. Clin Pharmacol Ther 2000; 67: 360 – 367.
  35. Paap CM, Nahata MC. Clinical pharmacokinetics of antibacterial drugs in neonates. Clin Pharamacokinet 1990; 19: 280–318.
  36. Volosov A, Soldin S J. Simultaneous measurement of 14 AIDS drugs by tandem-MS. Ther Drug Monit 2001; 23: 485.
  37. Back DJ, Khoo SH, Gibbons SE, Barry MG, Merry C. TDM of antiretrovirals in human immunodeficiency virus infection. Ther Drug Monit 2000; 22: 122–126.
  38. Vessell ES. On the significance of host factors that affect drug disposition. Clin Pharamcol Ther 1982; 31: 1–7.
  39. Dahl ML, Sjoqvist F. Pharamacogenetic methods as a compliment to therapeutic monitoring of antidepressant and neuroleptics. Ther Drug Monit 2000; 22: 114–117.
  40. Schmitz G, Aslanidis C, Lackner KJ. Pharmacogenomics: implications of laboratory medicine. Clin Chem Acta 2001; 308: 43–55.
  41. Evans WE, Mcleod HL. Pharmacogenomics-drug disposition, drug targets and side effects. N Engl J Med 2003; 348: 538–549.
  42. White R, Wong SHY. Pharmacogenomics and its clinical applications. MLO Med Lab Obs 2005; 37: 20–27.
  43. Feero WG, Guttmacher A E. Genomics, personalized medicine and pediatrics. Acad Pediatr 2014; 14(1): 14–22.
Share Button
(Visited 983 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.